Smo ablation in fibroblasts mitigates ischemic AKI
To elucidate the role of fibroblast-derived Smo in AKI, we generated fibroblast-specific Smo knockout mice by employing the tamoxifen-inducible Cre-LoxP system [9]. Briefly, homozygous Smo-floxed mice were mated with the Gli1-CreERT2 transgenic mice under the control of endogenous Gli1 promoter to create Smo fibroblast conditional knockout mice (genotype: Cre+/−, Smofl/fl; designated as Gli1-Smo-/-); Gli1 expression is confined to a subset of fibroblasts (Fig. 1A) [3, 9, 25–27]. Age and sex-matched Smo-floxed mice (genotype: Cre−/−, Smofl/fl; designated as Gli1-Smo+/+) from the same litters were used as controls. Gli1-Smo-/- mice were phenotypically normal. There was no appreciable abnormality in kidney function between Gli1-Smo-/- and Gli1-Smo+/+ mice (Supplementary Fig. 1A). After tamoxifen injections for five consecutive days, Cre-mediated recombination was induced in Gli1 + fibroblasts, as previously reported [9, 28, 29]. Mice were then subjected to bilateral renal ischemia-reperfusion injury (IRI) for 1 day to induce AKI after 1-week tamoxifen wash-out (Fig. 1A). Surprisingly, opposite to the results of loss of Shh in tubules,[4] deletion of Smo in Gli1 + fibroblasts preserved kidney function at 1 day after ischemic AKI. Compared to Gli1-Smo+/+ mice, serum creatinine levels were reduced by 22.7% in Gli1-Smo-/- mice (Fig. 1B). Of note, similar result was obtained in a separated ischemic AKI model for 3 days (data not shown). Consistently, Gli1-Smo-/- mice exhibited fewer AKI-associated morphologic changes, such as less cellular debris, brush border loss, and intratubular proteinaceous casts (Fig. 1C; Quantitative data shown in Fig. 1D). Furthermore, the expression of two classic acute tubular injury markers, kidney injury molecule 1 (Kim-1) and neutrophil gelatinase-associated lipocalin (NGAL) [30, 31], were also decreased in Gli1-Smo-/- mice at 1 day after ischemic AKI, compared to Gli1-Smo+/+ mice (Fig. 1E and 1F).
To confirm the above phenotypic changes, we assessed the differences in the key biological mechanisms involved in AKI development, inflammation and cell death [32, 33], between Gli1-Smo+/+ and Gli1-Smo-/- mice ischemic kidneys. Western blots revealed that tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) protein levels were significantly reduced in Gli1-Smo-/- mice, compared with Gli1-Smo+/+ mice (Fig. 1G and 1H). Accordingly, immunostaining results indicated fewer infiltrated CD45+ leukocytes, CD3+ T cells, and F4/80+ macrophages in Gli1-Smo-/- mice diseased kidneys (Fig. 1I, 1J, quantitative data in Fig. 1K, and Supplementary Fig. 1B).
Because AKI features sublethal or lethal damage of renal tubules [33], we further examined if loss of Smo in fibroblasts influences different types of tubular cell death in ischemic kidneys, such as apoptosis, necrosis, or ferroptosis [18], Western blots revealed that pro-apoptosis proteins FasL, Bad, and cleaved caspase-7 were reduced and anti-apoptosis protein Bcl-2 was increased in Gli1-Smo-/- mice, compared with Gli1-Smo+/+ mice [19] (Fig. 1L and 1M). Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) showed similar results (Fig. 1N and 1O). Intriguingly, neither ferroptosis marker Glutathione Peroxidase 4 (GPX4) nor necroptosis marker phosphorylated mixed lineage kinase domain-like protein (p-MLKL) was changed between Gli1-Smo+/+ and Gli1-Smo-/- kidneys after ischemic AKI (Supplementary Fig. 1C-1E). Collectively, these results suggested that ablation of Smo in fibroblasts protects against AKI.
Loss of Smo in Gli1 + fibroblasts enhances perivascular mesenchymal cell activity after AKI
Hh/Smo/Gli1 signaling is necessary for fibroblast proliferation in various diseases such as organ fibrogenesis, systemic sclerosis, and cancer microenvironment formation [23, 34–37]. We previously reported that early activated fibroblasts are indispensable for kidney repair after AKI [4]. Therefore, we were puzzled as to why loss of Gli1 + fibroblast-derived Smo alleviated AKI in our current model. Considering Gli1 + fibroblasts are a small fraction of the perivascular mesenchymal cell population [9, 28], we speculated that loss of Smo in Gli1 + fibroblasts may activate the remaining perivascular mesenchymal cells, such as Gli1- fibroblast and pericytes, by stimulating cell survival after AKI. To this end, we systemically assessed the expression of several markers for activated fibroblasts and pericytes in diseased kidneys, including fibroblast-specific protein 1 (FSP1), vimentin, α-smooth muscle actin (α-SMA), and platelet-derived growth factor receptor β (PDGFR-β). Quantitative real-time PCR (qRT-PCR) analyses revealed that mRNA expression of FSP1, vimentin, and α-SMA were significantly increased in Gli1-Smo-/- mice 1 day after ischemic AKI, compared with Gli1-Smo+/+ mice (Fig. 2A). Western blots further demonstrated marked elevation of PDGFR-β, vimentin, and α-SMA proteins in Gli1-Smo-/- mice (Fig. 2B; quantitative data are presented in Supplementary Fig. 2A). Consistently, immunostaining revealed increased expression of FSP-1, PDGFR-β, and vimentin in Gli1-Smo-/- mice diseased kidneys, especially in the juxtamedullary zone (Fig. 2C and Supplementary Fig. 2B-2D). To understand the distribution of proliferative cells in diseased kidneys after loss of Smo in Gli1 + fibroblasts, we assessed proliferating cell nuclear antigen (PCNA) and Ki67 levels. Western blots demonstrated an upregulation of PCNA in Gli1-Smo-/- mice at 1 day after ischemic AKI, compared with Gli1-Smo+/+ mice (Fig. 2B and Supplementary Fig. 2A). Immunohistochemical staining indicated that Ki67 + cells were mainly located in the interstitial compartment of Gli1-Smo-/- kidneys (Fig. 2D-2E). Together, these results indicated that ablation of Smo in fibroblasts in turn activated perivascular mesenchymal cells after ischemic AKI.
Global proteomics identifies nidogen-1 as a key participant in AKI repair after loss of Smo in fibroblasts
To better understand how loss of Smo in fibroblasts mitigates AKI, we used a label-free quantitative approach to profile the global proteome of Gli1-Smo+/+ and Gli1-Smo-/- mice kidneys at 1 day after IRI (Fig. 3A). Impressively, the principal component analysis (PCA) of our proteomic data clearly classified Gli1-Smo+/+ and Gli1-Smo-/- mice according to their genotype (Fig. 3B). A multiple-variation test by ANOVA identified 680 proteins with significantly different expression (Permutation FDR 0.05) between Gli1-Smo+/+ and Gli1-Smo-/- mice diseased kidneys (Quantified proteins are presented in Supplementary Table 1). Compared to Gli1-Smo+/+ mice, 206 and 346 proteins were up- and down-regulated respectively in Gli1-Smo-/- mice diseased kidneys after ischemic AKI (Fig. 3C and Supplementary Table 2; Correlations between biological replicates within the same group and the distribution of protein intensity are presented in Supplementary Fig. 3A-3B). The following Gene Ontology (GO) cellular compartment analysis revealed that these proteins were generally distributed in mitochondrion, extracellular exosome, peroxisome, cytosol, and ECM (Fig. 3D). The activation of perivascular mesenchymal cells in Gli1-Smo-/- mice is redolent of enhanced ECM synthesis in diseased kidneys (Fig. 2). Indeed, unlike most proteins that showed down-regulated trends in other cellular compartments, 110 out of 141 ECM proteins were upregulated in Gli1-Smo-/- mice compared with Gli1-Smo+/+ mice (Fig. 3D). This phenomenon indicates that ECM remodeling by enhanced protein synthesis may play a critical role in Gli1-Smo-/- kidney repair.
We therefore focused on the activated ECM proteins with significant up-regulations in Gli1-Smo-/- mice kidneys. To select the most significantly impacted ECM protein for further investigation, we excluded those that have been well studied [such as vimentin (Fig. 2A-2C) and prelamin-A/C]. Among the remaining proteins, we paid our attention to nidogen-1 (NID1), one of the most prominent matrix proteins upregulated in Gli1-Smo-/- mice (Fig. 3E-3F and Supplementary Fig. 3C) which has been reported as an essential component of the basement membrane that plays a role in cell interactions with the ECM. Since ECM is identified as a major cellular compartment of proteins distributed, we hypothesized that ECM could organize a favorable kidney local microenvironment to repair kidneys after AKI. Thus, we chose NID1 for further study.
To confirm our findings, we assessed NID1 levels in Gli1-Smo+/+ and Gli1-Smo-/- mice diseased kidneys. qPCR analysis indicated that NID1 mRNA level has an upregulation trend (albeit statistically insignificant) in Gli1-Smo-/- mice as compare to Gli1-Smo+/+ (Fig. 3G), and western blots demonstrated a marked elevation of NID1 protein in Gli1-Smo-/- mice (Fig. 3H and Supplementary Fig. 3D), compared with Gli1-Smo+/+ mice. In a separate analysis using single nucleus RNA sequencing, NID1 was highly expressed in fibroblasts and pericytes after IRI at 12 hours (Fig. 3I) [38]. Consistently, immunohistochemical staining confirmed enhanced NID1 distribution in the interstitium of Gli1-Smo-/- ischemic kidneys (Fig. 3J). To further establish the molecular link between NID1 and Smo, we identified multiple potential binding sites of NID1 and Smo in the optimal conformation at pose 51 through virtual protein-protein ZRDOCK and RDOCK algorithms (Supplementary Fig. 4). Immunoprecipitation of Smo or NID1 pulled down NID1 or Smo, respectively, which indicating Smo physically binds to NID1 (Fig. 3K). In addition, to obtain a broader view of the dysregulated pathways after loss of Smo in fibroblasts, we constructed a signaling network based on the STRING database. The analysis with the highest confidence score (cutoff = 0.9) resulted in 413 nodes and 1815 interactions. (Supplementary Fig. 3E). The majority of interactions and nodes were associated with Wnt, Hh, and PPAR-γ signaling pathways and lipid metabolism. These data suggested that loss of Smo in Gli1 + fibroblasts liberated NID1, allowing it to orchestrate a favorable matrix microenvironment after AKI.
Phosphoproteomics reveals that Smo deletion in fibroblasts remodels the Wnt signaling pathway
To gain more insights into the temporal regulation and functional changes in signaling of Smo-deficient fibroblasts in AKI repair, we performed phosphoproteomics on the same set of kidney samples used for the global proteomics (Fig. 4A). Similar to the global proteomics, PCA of the phosphoproteome clearly separated Gli1-Smo+/+ and Gli1-Smo-/- mice kidneys (Fig. 4B). Impressively, most of the significantly different phosphopeptides were decreased in Gli1-Smo-/- mice compared with Gli1-Smo+/+ kidneys (Supplementary Table 3). We then filtered the phosphorylation data set to include those phosphopeptides that were quantified in at least three replicates and used t-test analysis with p-value correnction (Permutation FDR 0.05). Enrichment analysis indicated that phosphoproteins associated with RNA splicing, cell-cell adhesion, and mRNA processing were significantly over-represented (p < 10− 5). Consistent with network analysis performed with global proteomics (Supplementary Fig. 3E), a clear reduced phosphorylating of multiple components of the Wnt signaling pathway in Gli1-Smo-/- mice was observed (Supplementary Fig. 5A). Pairwise comparisons between Gli1-Smo+/+ and Gli1-Smo-/- mice at 1 day after IRI highlighted significantly changed Wnt components, such as pGSK3β, pSlc9a3r1, pCtnnd1, pTGFβ1i1, pCcny, and pLeo1 (Fig. 4C and Supplementary Fig. 5B-5C). In particular, we identified two downregulated tyrosine phosphorylation sites (Tyr216 and Tyr279) on GSK-3β in Gli1-Smo-/- mice. Dephosphorylation on Tyr216 and Tyr279 represses the activity of GSK-3β. Therefore, in Gli1-Smo-/- mice, inactivated GSK-3β loses its capacity to destabilize β-catenin, a principal protein in the Wnt signaling pathway. This ultimately causes the activation of the canonical Wnt/β-catenin signaling pathway. In addition, the network analysis showed that GSK-3β interacts with other proteins in Gli1-Smo-/- kidneys (Supplementary Fig. 5B).
Previously, we and others reported that activation of the Wnt signaling pathway protected against AKI [8, 10, 12, 39]. Based on the information provided by the phosphoproteome analysis, we assessed the status of the canonical Wnt signaling pathway in Gli1-Smo+/+ and Gli1-Smo-/- mice after AKI. qRT-PCR revealed that 10 out of 19 Wnt family members (Wnt 1, 2, 4, 5A, 5B, 7A, 7B, 9B, 10A, and 11) were significantly induced in Gli1-Smo-/- mice (Fig. 4D). In the same tissue, several Frizzled receptors in the Wnt signaling pathway were also upregulated in Gli1-Smo-/- mice (Supplementary Fig. 6A). Western blots demonstrated that β-catenin, Wnt 1, and Wnt 5A/B proteins in the Wnt pathway were markedly induced in Gli1-Smo-/- mice (Fig. 4E; quantitative data are presented in Supplementary Fig. 6B). Immunohistochemical staining confirmed increased expression of β-catenin, Wnt1, Wnt4, and Wnt5A/B in Gli1-Smo-/- mice diseased tubular cells, compared with Gli1-Smo+/+ mice (Fig. 4F). Of particular interest, three Hh ligands were also induced in Gli1-Smo-/- mice after AKI (Supplementary Fig. 7), although the mechanisms involved remain unclear. Taken together, our data suggest that ablation of Smo in fibroblasts is linked to Wnt signaling pathway activation after ischemic AKI.
Pharmaceutical inhibition of Smo in fibroblasts promotes tubular cell survival through NID1
To further decipher the role of fibroblast-derived Smo in regulating tubular cell survival, we treated normal rat kidney fibroblasts (NRK-49F) with cyclopamine (CPN), a small-molecule, Smo-specific inhibitor. Under hypoxia stress induced by CoCl2, blocking Smo activity significantly promoted the expression of PDGFR-β, vimentin, α-SMA, and PCNA proteins in cultured fibroblasts (Fig. 5A). NID1 protein was also induced by CoCl2 and CPN in fibroblasts (Fig. 5B), and immunofluorescence staining of whole cells showed similar results (Fig. 5C).
To validate our in vivo findings that loss of fibroblast-selective Smo induced tubular Wnt signals and reduced cell apoptosis, we performed several in vitro and ex vivo experiments. For our in vitro experiment, we treated normal rat kidney epithelial cells (NRK-52E) with NID1-enriched conditioned medium (CM) collected from fibroblasts (Fig. 5D). Under basal conditions, NID1-enriched CM did not cause dramatic changes in the expression of β-catenin, Wnt1, Wnt2, and Wnt5A/B in NRK-52E cells (Fig. 5E). However, these proteins were significantly induced under hypoxic stress after incubated with NID1-enriched CM (Fig. 5F). Besides NID1-enriched CM, we further applied NID1 recombinant protein to treat NRK-52E at different dosages, as illustrated in Fig. 5D. In this experiment, NID1 activated β-catenin, Wnt1, Wnt5A/B, and Wnt16 in both basal and hypoxic conditions (Fig. 5G and 5H). Impressively, either NID1-enriched CM or NID1 recombinant protein possessed the capacity to reduce tubular cell apoptosis. Specifically, less caspase 3 cleavage was observed in NRK-52E cells after treatment with NID1-enriched CM or NID1 recombinant protein and stimulation with a classic apoptosis inducer, staurosporine (Fig. 5I and 5J). Immunofluorescence staining for cleaved caspase-3 confirmed these findings (Fig. 5K-5N).
To better mimic the in vivo microenvironment, we then isolated decellularized fibroblast scaffolds after hypoxia stress for 24 hours and seeded NRK-52E cells on the scaffold-coated plate (Fig. 6A; as we used in a previous study [40]). After treatment with CPN, NID1 was significantly enriched in the decellularized fibroblast matrix scaffold (Fig. 6B). Similarly, under basal conditions, there were minor differences of β-catenin, Wnt 2, and Wnt 5A/B expression in the seeded tubular cells between control scaffolds and NID1-enriched scaffolds (Fig. 6C). Under hypoxic stress, the NID1-enriched scaffold exhibited enhanced capacity to activate β-catenin, Wnt 2, and Wnt 5A/B and repress caspase-3 cleavage in NRK-52E cells (Fig. 6D-6E). Our results suggest that inhibiting Smo in fibroblasts liberated NID1 to activate tubular Wnt signaling to protect against AKI (Fig. 6F).